Radiosensitizing Hypoxic Tumour Cells: Comparison
Please note this is a comparison between Version 2 by Conner Chen and Version 1 by Alyssa Apilan.

One of the most difficult barriers encountered when treating the majority of solid tumours is attributed to the scattered microregions within the tumour characterized by the lack of oxygen. This is known as tumour hypoxia. The lack of oxygen supply results in hypoxic microregions scattered throughout the tumour relative to normal tissue. The hypoxic environments within tumours create several cancer treatment barriers. Most notably, all hypoxic cells are resistant to ionizing radiation (IR). The mechanism by which radiation is able to eradicate tumour cells by damaging DNA, resulting in apoptosis and cell death, occurs through the production of reactive oxygen species (ROS). However, due to the limited oxygen availability within hypoxic tumour microenvironments, this impedes the efficacy of radiotherapy. Furthermore, normal tissues are unable to withstand increased doses of radiation that compensate for tumour hypoxia. Methods targeting tumour hypoxia through radiosensitizing hypoxic cells include hyperbaric oxygen, hypoxic cell radiosensitizers, hypoxic cytotoxins, and tumour metabolism. As research continues to elucidate the relationship between tumour hypoxia and radiotherapy, novel approaches have been developed.

  • hypoxia
  • non-targeted effects
  • autophagy
  • PET imaging
  • radiosensitizers
Please wait, diff process is still running!

References

  1. Ogawa, K.; Kohshi, K.; Ishiuchi, S.; Matsushita, M.; Yoshimi, N.; Murayama, S. Old but New Methods in Radiation Oncology: Hyperbaric Oxygen Therapy. Int. J. Clin. Oncol. 2013, 18, 364–370.
  2. Jain, K.K. Textbook of Hyperbaric Medicine; Springer International Publishing: Cham, Switzerland, 1953; ISBN 978-3-319-47138-9.
  3. Hartmann, K.A.; van der Kleij, A.J.; Carl, U.M.; Hulshof, M.C.; Willers, R.; Sminia, P. Effects of Hyperbaric Oxygen and Normobaric Carbogen on the Radiation Response of the Rat Rhabdomyosarcoma R1H. Int. J. Radiat. Oncol. Biol. Phys. 2001, 51, 1037–1044.
  4. Hall, E.J.; Giaccia, A.J. Radiobiology for the Radiologist, 7th ed.; Wolters Kluwer Health/Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2012; ISBN 978-1-60831-193-4.
  5. Churchill-Davidson, I. The Oxygen Effect in Radiotherapy—Historical Review. Hyperb. Oxyg. Radiat. Ther. Cancer 1968, 1, 1–15.
  6. Bennett, M.H.; Feldmeier, J.; Smee, R.; Milross, C. Hyperbaric Oxygenation for Tumour Sensitisation to Radiotherapy. Cochrane Database Syst. Rev. 2018, 2018, CD005007.
  7. Kohshi, K.; Kinoshita, Y.; Imada, H.; Kunugita, N.; Abe, H.; Terashima, H.; Tokui, N.; Uemura, S. Effects of Radiotherapy after Hyperbaric Oxygenation on Malignant Gliomas. Br. J. Cancer 1999, 80, 236–241.
  8. Moen, I.; Stuhr, L.E.B. Hyperbaric Oxygen Therapy and Cancer—A Review. Target. Oncol. 2012, 7, 233–242.
  9. Kjellen, E.; Joiner, M.C.; Collier, J.M.; Johns, H.; Rojas, A. A Therapeutic Benefit from Combining Normobaric Carbogen or Oxygen with Nicotinamide in Fractionated X-Ray Treatments. Radiother. Oncol. J. Eur. Soc. Ther. Radiol. Oncol. 1991, 22, 81–91.
  10. Tran, L.-B.-A.; Bol, A.; Labar, D.; Karroum, O.; Bol, V.; Jordan, B.; Grégoire, V.; Gallez, B. Potential Role of Hypoxia Imaging Using 18F-FAZA PET to Guide Hypoxia-Driven Interventions (Carbogen Breathing or Dose Escalation) in Radiation Therapy. Radiother. Oncol. 2014, 113, 204–209.
  11. Harrison, L.B.; Chadha, M.; Hill, R.J.; Hu, K.; Shasha, D. Impact of Tumour Hypoxia and Anemia on Radiation Therapy Outcomes. Oncologist 2002, 7, 492–508.
  12. Fleming, I.N.; Manavaki, R.; Blower, P.J.; West, C.; Williams, K.J.; Harris, A.L.; Domarkas, J.; Lord, S.; Baldry, C.; Gilbert, F.J. Imaging Tumour Hypoxia with Positron Emission Tomography. Br. J. Cancer 2015, 112, 238–250.
  13. Unterrainer, M.; Eze, C.; Ilhan, H.; Marschner, S.; Roengvoraphoj, O.; Schmidt-Hegemann, N.S.; Walter, F.; Kunz, W.G.; af Rosenschöld, P.M.; Jeraj, R.; et al. Recent Advances of PET Imaging in Clinical Radiation Oncology. Radiat. Oncol. 2020, 15, 88.
  14. Chapman, J.D. Hypoxic Sensitizers—Implications for Radiation Therapy. N. Engl. J. Med. 1979, 301, 1429–1432.
  15. Nunn, A.; Linder, K.; Strauss, H.W. Nitroimidazoles and Imaging Hypoxia. Eur. J. Nucl. Med. 1995, 22, 265–280.
  16. Edwards, D.I. Nitroimidazole Drugs--Action and Resistance Mechanisms. I. Mechanisms of Action. J. Antimicrob. Chemother. 1993, 31, 9–20.
  17. Lopci, E.; Grassi, I.; Chiti, A.; Nanni, C.; Cicoria, G.; Toschi, L.; Fonti, C.; Lodi, F.; Mattioli, S.; Fanti, S. PET Radiopharmaceuticals for Imaging of Tumour Hypoxia: A Review of the Evidence. Am. J. Nucl. Med. Mol. Imaging 2014, 4, 365–384.
  18. Bourgeois, M.; Rajerison, H.; Guerard, F.; Mougin-Degraef, M.; Barbet, J.; Michel, N.; Cherel, M.; Faivre-Chauvet, A. Contribution of [64Cu]-ATSM PET in Molecular Imaging of Tumour Hypoxia Compared to Classical [18F]-MISO—A Selected Review. Nucl. Med. Rev. Cent. East. Eur. 2011, 14, 90–95.
  19. Hoigebazar, L.; Jeong, J.M. Hypoxia Imaging Agents Labeled with Positron Emitters. Recent Results Cancer Res. Fortschr. Krebsforsch. Prog. Dans Rech. Sur Cancer 2013, 194, 285–299.
  20. Takasawa, M.; Moustafa, R.R.; Baron, J.-C. Applications of Nitroimidazole in Vivo Hypoxia Imaging in Ischemic Stroke. Stroke 2008, 39, 1629–1637.
  21. Kumar, P.; Stypinski, D.; Xia, H.; McEwan, A.J.B.; Machulla, H.-J.; Wiebe, L.I. Fluoroazomycin Arabinoside (FAZA): Synthesis, 2H and 3H-Labelling and Preliminary Biological Evaluation of a Novel 2-Nitroimidazole Marker of Tissue Hypoxia. J. Label. Compd. Radiopharm. 1999, 42, 3–16.
  22. Busk, M.; Horsman, M.R.; Jakobsen, S.; Bussink, J.; van der Kogel, A.; Overgaard, J. Cellular Uptake of PET Tracers of Glucose Metabolism and Hypoxia and Their Linkage. Eur. J. Nucl. Med. Mol. Imaging 2008, 35, 2294–2303.
  23. Quartuccio, N.; Laudicella, R.; Mapelli, P.; Guglielmo, P.; Pizzuto, D.A.; Boero, M.; Arnone, G.; Picchio, M. Young AIMN Working Group. Hypoxia PET Imaging beyond 18F-FMISO in Patients with High-Grade Glioma: 18F-FAZA and Other Hypoxia Radiotracers. Clin. Transl. Imaging 2020, 8, 11–20.
  24. Busk, M.; Mortensen, L.S.; Nordsmark, M.; Overgaard, J.; Jakobsen, S.; Hansen, K.V.; Theil, J.; Kallehauge, J.F.; D’Andrea, F.P.; Steiniche, T.; et al. PET Hypoxia Imaging with FAZA: Reproducibility at Baseline and during Fractionated Radiotherapy in Tumour-Bearing Mice. Eur. J. Nucl. Med. Mol. Imaging 2013, 40, 186–197.
  25. Adams, G.E.; Flockhart, I.R.; Smithen, C.E.; Stratford, I.J.; Wardman, P.; Watts, M.E. Electron-Affinic Sensitization. VII. A Correlation between Structures, One-Electron Reduction Potentials, and Efficiencies of Nitroimidazoles as Hypoxic Cell Radiosensitizers. Radiat. Res. 1976, 67, 9–20.
  26. Wardman, P. Chemical Radiosensitizers for Use in Radiotherapy. Clin. Oncol. 2007, 19, 397–417.
  27. Hall, E.J. Radiobiology for the Radiologist, 5th ed.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2000; ISBN 978-0-7817-2649-8.
  28. Overgaard, J.; Sand Hansen, H.; Lindeløv, B.; Overgaard, M.; Jørgensen, K.; Rasmusson, B.; Berthelsen, A. Nimorazole as a Hypoxic Radiosensitizer in the Treatment of Supraglottic Larynx and Pharynx Carcinoma. First Report from the Danish Head and Neck Cancer Study (DAHANCA) Protocol 5-85. Radiother. Oncol. J. Eur. Soc. Ther. Radiol. Oncol. 1991, 20 (Suppl. S1), 143–149.
  29. Overgaard, J.; Hansen, H.S.; Overgaard, M.; Bastholt, L.; Berthelsen, A.; Specht, L.; Lindeløv, B.; Jørgensen, K. A Randomized Double-Blind Phase III Study of Nimorazole as a Hypoxic Radiosensitizer of Primary Radiotherapy in Supraglottic Larynx and Pharynx Carcinoma. Results of the Danish Head and Neck Cancer Study (DAHANCA) Protocol 5-85. Radiother. Oncol. J. Eur. Soc. Ther. Radiol. Oncol. 1998, 46, 135–146.
  30. Graham, K.; Unger, E. Overcoming Tumour Hypoxia as a Barrier to Radiotherapy, Chemotherapy and Immunotherapy in Cancer Treatment. Int. J. Nanomed. 2018, 13, 6049–6058.
  31. Teicher, B.A.; Lazo, J.S.; Sartorelli, A.C. Classification of Antineoplastic Agents by Their Selective Toxicities toward Oxygenated and Hypoxic Tumour Cells. Cancer Res. 1981, 41, 73–81.
  32. Wardman, P. Nitroimidazoles as Hypoxic Cell Radiosensitizers and Hypoxia Probes: Misonidazole, Myths and Mistakes. Br. J. Radiol. 2019, 92, 20170915.
  33. Wang, H.; Mu, X.; He, H.; Zhang, X.-D. Cancer Radiosensitizers. Trends Pharmacol. Sci. 2018, 39, 24–48.
  34. Matsumura, Y.; Maeda, H. A New Concept for Macromolecular Therapeutics in Cancer Chemotherapy: Mechanism of Tumouritropic Accumulation of Proteins and the Antitumour Agent Smancs. Cancer Res. 1986, 46, 6387–6392.
  35. Wong, A.D.; Ye, M.; Ulmschneider, M.B.; Searson, P.C. Quantitative Analysis of the Enhanced Permeation and Retention (EPR) Effect. PLoS ONE 2015, 10, e0123461.
  36. Minchinton, A.I.; Tannock, I.F. Drug Penetration in Solid Tumours. Nat. Rev. Cancer 2006, 6, 583–592.
  37. Golombek, S.K.; May, J.-N.; Theek, B.; Appold, L.; Drude, N.; Kiessling, F.; Lammers, T. Tumour Targeting via EPR: Strategies to Enhance Patient Responses. Adv. Drug Deliv. Rev. 2018, 130, 17–38.
  38. Torchilin, V. Tumour Delivery of Macromolecular Drugs Based on the EPR Effect. Adv. Drug Deliv. Rev. 2011, 63, 131–135.
  39. Prabhakar, U.; Maeda, H.; Jain, R.K.; Sevick-Muraca, E.M.; Zamboni, W.; Farokhzad, O.C.; Barry, S.T.; Gabizon, A.; Grodzinski, P.; Blakey, D.C. Challenges and Key Considerations of the Enhanced Permeability and Retention (EPR) Effect for Nanomedicine Drug Delivery in Oncology. Cancer Res. 2013, 73, 2412–2417.
  40. Maeda, H.; Nakamura, H.; Fang, J. The EPR Effect for Macromolecular Drug Delivery to Solid Tumours: Improvement of Tumour Uptake, Lowering of Systemic Toxicity, and Distinct Tumour Imaging in Vivo. Adv. Drug Deliv. Rev. 2013, 65, 71–79.
  41. Hashizume, H.; Baluk, P.; Morikawa, S.; McLean, J.W.; Thurston, G.; Roberge, S.; Jain, R.K.; McDonald, D.M. Openings between Defective Endothelial Cells Explain Tumour Vessel Leakiness. Am. J. Pathol. 2000, 156, 1363–1380.
  42. Peer, D.; Karp, J.M.; Hong, S.; Farokhzad, O.C.; Margalit, R.; Langer, R. Nanocarriers as an Emerging Platform for Cancer Therapy. Nat. Nanotechnol. 2007, 2, 751–760.
  43. Cheng, Z.; Al Zaki, A.; Hui, J.Z.; Muzykantov, V.R.; Tsourkas, A. Multifunctional Nanoparticles: Cost versus Benefit of Adding Targeting and Imaging Capabilities. Science 2012, 338, 903–910.
  44. Her, S.; Jaffray, D.A.; Allen, C. Gold Nanoparticles for Applications in Cancer Radiotherapy: Mechanisms and Recent Advancements. Adv. Drug Deliv. Rev. 2017, 109, 84–101.
  45. Mi, Y.; Shao, Z.; Vang, J.; Kaidar-Person, O.; Wang, A.Z. Application of Nanotechnology to Cancer Radiotherapy. Cancer Nanotechnol. 2016, 7, 11.
  46. Babaei, M.; Ganjalikhani, M. The Potential Effectiveness of Nanoparticles as Radio Sensitizers for Radiotherapy. BioImpacts BI 2014, 4, 15–20.
  47. Chen, Y.; Yang, J.; Fu, S.; Wu, J. Gold Nanoparticles as Radiosensitizers in Cancer Radiotherapy. Int. J. Nanomed. 2020, 15, 9407–9430.
  48. Dimitriou, N.M.; Tsekenis, G.; Balanikas, E.C.; Pavlopoulou, A.; Mitsiogianni, M.; Mantso, T.; Pashos, G.; Boudouvis, A.G.; Lykakis, I.N.; Tsigaridas, G.; et al. Gold Nanoparticles, Radiations and the Immune System: Current Insights into the Physical Mechanisms and the Biological Interactions of This New Alliance towards Cancer Therapy. Pharmacol. Ther. 2017, 178, 1–17.
  49. Retif, P.; Pinel, S.; Toussaint, M.; Frochot, C.; Chouikrat, R.; Bastogne, T.; Barberi-Heyob, M. Nanoparticles for Radiation Therapy Enhancement: The Key Parameters. Theranostics 2015, 5, 1030–1044.
  50. Yao, X.; Huang, C.; Chen, X.; Yi, Z.; Sanche, L. Chemical Radiosensitivity of DNA Induced by Gold Nanoparticles. J. Biomed. Nanotechnol. 2015, 11, 478–485.
  51. Liu, K.; Han, L.; Zhuang, J.; Yang, D.-P. Protein-Directed Gold Nanoparticles with Excellent Catalytic Activity for 4-Nitrophenol Reduction. Mater. Sci. Eng. C Mater. Biol. Appl. 2017, 78, 429–434.
  52. Mateo, D.; Morales, P.; Ávalos, A.; Haza, A.I. Oxidative Stress Contributes to Gold Nanoparticle-Induced Cytotoxicity in Human Tumour Cells. Toxicol. Mech. Methods 2014, 24, 161–172.
  53. Decrock, E.; Hoorelbeke, D.; Ramadan, R.; Delvaeye, T.; De Bock, M.; Wang, N.; Krysko, D.V.; Baatout, S.; Bultynck, G.; Aerts, A.; et al. Calcium, Oxidative Stress and Connexin Channels, a Harmonious Orchestra Directing the Response to Radiotherapy Treatment? Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 1099–1120.
  54. Sinclair, W.K.; Morton, R.A. X-Ray Sensitivity during the Cell Generation Cycle of Cultured Chinese Hamster Cells. Radiat. Res. 1966, 29, 450–474.
  55. Mackey, M.A.; Saira, F.; Mahmoud, M.A.; El-Sayed, M.A. Inducing Cancer Cell Death by Targeting Its Nucleus: Solid Gold Nanospheres versus Hollow Gold Nanocages. Bioconjug. Chem. 2013, 24, 897–906.
  56. Cui, L.; Tse, K.; Zahedi, P.; Harding, S.M.; Zafarana, G.; Jaffray, D.A.; Bristow, R.G.; Allen, C. Hypoxia and Cellular Localization Influence the Radiosensitizing Effect of Gold Nanoparticles (AuNPs) in Breast Cancer Cells. Radiat. Res. 2014, 182, 475–488.
  57. Pan, Y.; Leifert, A.; Ruau, D.; Neuss, S.; Bornemann, J.; Schmid, G.; Brandau, W.; Simon, U.; Jahnen-Dechent, W. Gold Nanoparticles of Diameter 1.4 Nm Trigger Necrosis by Oxidative Stress and Mitochondrial Damage. Small 2009, 5, 2067–2076.
  58. Butterworth, K.T.; Coulter, J.A.; Jain, S.; Forker, J.; McMahon, S.J.; Schettino, G.; Prise, K.M.; Currell, F.J.; Hirst, D.G. Evaluation of Cytotoxicity and Radiation Enhancement Using 1.9 Nm Gold Particles: Potential Application for Cancer Therapy. Nanotechnology 2010, 21, 295101.
  59. Liu, Y.; Zhang, P.; Li, F.; Jin, X.; Li, J.; Chen, W.; Li, Q. Metal-Based NanoEnhancers for Future Radiotherapy: Radiosensitizing and Synergistic Effects on Tumour Cells. Theranostics 2018, 8, 1824–1849.
  60. Djuzenova, C.S.; Elsner, I.; Katzer, A.; Worschech, E.; Distel, L.V.; Flentje, M.; Polat, B. Radiosensitivity in Breast Cancer Assessed by the Histone γ-H2AX and 53BP1 Foci. Radiat. Oncol. 2013, 8, 98.
  61. Vignard, J.; Mirey, G.; Salles, B. Ionizing-Radiation Induced DNA Double-Strand Breaks: A Direct and Indirect Lighting Up. Radiother. Oncol. J. Eur. Soc. Ther. Radiol. Oncol. 2013, 108, 362–369.
  62. Mothersill, C.; Crean, M.; Lyons, M.; McSweeney, J.; Mooney, R.; O’Reilly, J.; Seymour, C.B. Expression of Delayed Toxicity and Lethal Mutations in the Progeny of Human Cells Surviving Exposure to Radiation and other Environmental Mutagens. Int. J. Radiat. Biol. 1998, 74, 673–680.
  63. Glaviano, A.; Nayak, V.; Cabuy, E.; Baird, D.M.; Yin, Z.; Newson, R.; Ladon, D.; Rubio, M.A.; Slijepcevic, P.; Lyng, F.; et al. Effects of HTERT on Metal Ion-Induced Genomic Instability. Oncogene 2006, 25, 3424–3435.
  64. Glaviano, A.; Mothersill, C.; Case, C.P.; Rubio, M.A.; Newson, R.; Lyng, F. Effects of HTERT on Genomic Instability Caused by Either Metal or Radiation or Combined Exposure. Mutagenesis 2009, 24, 25–33.
  65. Coen, N.; Kadhim, M.A.; Wright, E.G.; Case, C.P.; Mothersill, C.E. Particulate Debris from a Titanium Metal Prosthesis Induces Genomic Instability in Primary Human Fibroblast Cells. Br. J. Cancer 2003, 88, 548–552.
  66. Rosa, S.; Connolly, C.; Schettino, G.; Butterworth, K.T.; Prise, K.M. Biological Mechanisms of Gold Nanoparticle Radiosensitization. Cancer Nanotechnol. 2017, 8, 2.
  67. Ghita, M.; McMahon, S.J.; Taggart, L.E.; Butterworth, K.T.; Schettino, G.; Prise, K.M. A Mechanistic Study of Gold Nanoparticle Radiosensitisation Using Targeted Microbeam Irradiation. Sci. Rep. 2017, 7, 44752.
  68. Brown, J.M.; Siim, B.G. Hypoxia-Specific Cytotoxins in Cancer Therapy. Semin. Radiat. Oncol. 1996, 6, 22–36.
  69. Zeng, Y.; Ma, J.; Zhan, Y.; Xu, X.; Zeng, Q.; Liang, J.; Chen, X. Hypoxia-Activated Prodrugs and Redox-Responsive Nanocarriers. Int. J. Nanomed. 2018, 13, 6551–6574.
  70. Rischin, D.; Peters, L.; Fisher, R.; Macann, A.; Denham, J.; Poulsen, M.; Jackson, M.; Kenny, L.; Penniment, M.; Corry, J.; et al. Tirapazamine, Cisplatin, and Radiation versus Fluorouracil, Cisplatin, and Radiation in Patients with Locally Advanced Head and Neck Cancer: A Randomized Phase II Trial of the Trans-Tasman Radiation Oncology Group (TROG 98.02). J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2005, 23, 79–87.
  71. Thambi, T.; Park, J.H.; Lee, D.S. Hypoxia-Responsive Nanocarriers for Cancer Imaging and Therapy: Recent Approaches and Future Perspectives. Chem. Commun. 2016, 52, 8492–8500.
  72. Gao, G.H.; Li, Y.; Lee, D.S. Environmental PH-Sensitive Polymeric Micelles for Cancer Diagnosis and Targeted Therapy. J. Control. Release 2013, 169, 180–184.
  73. Visser, J.G.; Van Staden, A.D.P.; Smith, C. Harnessing Macrophages for Controlled-Release Drug Delivery: Lessons From Microbes. Front. Pharmacol. 2019, 10, 22.
  74. Yu, H.; Yang, Z.; Li, F.; Xu, L.; Sun, Y. Cell-Mediated Targeting Drugs Delivery Systems. Drug Deliv. 2020, 27, 1425–1437.
  75. Dong, X.; Chu, D.; Wang, Z. Leukocyte-Mediated Delivery of Nanotherapeutics in Inflammatory and Tumour Sites. Theranostics 2017, 7, 751–763.
  76. Pierigè, F.; Serafini, S.; Rossi, L.; Magnani, M. Cell-Based Drug Delivery. Adv. Drug Deliv. Rev. 2008, 60, 286–295.
  77. Yousefpour, P.; Chilkoti, A. Co-Opting Biology to Deliver Drugs. Biotechnol. Bioeng. 2014, 111, 1699–1716.
  78. Wang, H.-F.; Liu, Y.; Yang, G.; Zhao, C.-X. Macrophage-Mediated Cancer Drug Delivery. Mater. Today Sustain. 2021, 11–12, 100055.
  79. Liu, X.; Li, J.; Peng, X.; Lv, B.; Wang, P.; Zhao, X.; Yu, B. Geraniin Inhibits LPS-Induced THP-1 Macrophages Switching to M1 Phenotype via SOCS1/NF-ΚB Pathway. Inflammation 2016, 39, 1421–1433.
  80. Zhu, Y.; Li, X.; Chen, J.; Chen, T.; Shi, Z.; Lei, M.; Zhang, Y.; Bai, P.; Li, Y.; Fei, X. The Pentacyclic Triterpene Lupeol Switches M1 Macrophages to M2 and Ameliorates Experimental Inflammatory Bowel Disease. Int. Immunopharmacol. 2016, 30, 74–84.
  81. Ponnaiya, B.; Cornforth, M.N.; Ullrich, R.L. Radiation-Induced Chromosomal Instability in BALB/c and C57BL/6 Mice: The Difference Is as Clear as Black and White. Radiat. Res. 1997, 147, 121–125.
  82. Mothersill, C.; Rea, D.; Wright, E.G.; Lorimore, S.A.; Murphy, D.; Seymour, C.B.; O’Malley, K. Individual Variation in the Production of a “bystander Signal” Following Irradiation of Primary Cultures of Normal Human Urothelium. Carcinogenesis 2001, 22, 1465–1471.
  83. Lindsay, K.J.; Coates, P.J.; Lorimore, S.A.; Wright, E.G. The Genetic Basis of Tissue Responses to Ionizing Radiation. Br. J. Radiol. 2007, 80, S2–S6.
  84. Lorimore, S.A.; Mukherjee, D.; Robinson, J.I.; Chrystal, J.A.; Wright, E.G. Long-Lived Inflammatory Signaling in Irradiated Bone Marrow Is Genome Dependent. Cancer Res. 2011, 71, 6485–6491.
  85. Mukherjee, D.; Coates, P.J.; Lorimore, S.A.; Wright, E.G. Responses to Ionizing Radiation Mediated by Inflammatory Mechanisms. J. Pathol. 2014, 232, 289–299.
  86. Poh, A.R.; Ernst, M. Targeting Macrophages in Cancer: From Bench to Bedside. Front. Oncol. 2018, 8, 49.
  87. Chanmee, T.; Ontong, P.; Konno, K.; Itano, N. Tumour-Associated Macrophages as Major Players in the Tumour Microenvironment. Cancers 2014, 6, 1670–1690.
  88. Aras, S.; Zaidi, M.R. TAMeless Traitors: Macrophages in Cancer Progression and Metastasis. Br. J. Cancer 2017, 117, 1583–1591.
  89. Zhang, M.; He, Y.; Sun, X.; Li, Q.; Wang, W.; Zhao, A.; Di, W. A High M1/M2 Ratio of Tumour-Associated Macrophages Is Associated with Extended Survival in Ovarian Cancer Patients. J. Ovarian Res. 2014, 7, 19.
  90. Gustafson, H.H.; Holt-Casper, D.; Grainger, D.W.; Ghandehari, H. Nanoparticle Uptake: The Phagocyte Problem. Nano Today 2015, 10, 487–510.
  91. Batrakova, E.V.; Gendelman, H.E.; Kabanov, A.V. Cell-Mediated Drugs Delivery. Expert Opin. Drug Deliv. 2011, 8, 415–433.
  92. Nowacek, A.S.; Miller, R.L.; McMillan, J.; Kanmogne, G.; Kanmogne, M.; Mosley, R.L.; Ma, Z.; Graham, S.; Chaubal, M.; Werling, J.; et al. NanoART Synthesis, Characterization, Uptake, Release and Toxicology for Human Monocyte-Macrophage Drug Delivery. Nanomedicine 2009, 4, 903–917.
  93. Söllner, T.; Bennett, M.K.; Whiteheart, S.W.; Scheller, R.H.; Rothman, J.E. A Protein Assembly-Disassembly Pathway in Vitro That May Correspond to Sequential Steps of Synaptic Vesicle Docking, Activation, and Fusion. Cell 1993, 75, 409–418.
  94. Moriwaki, T.; Okamoto, S.; Sasanuma, H.; Nagasawa, H.; Takeda, S.; Masunaga, S.-I.; Tano, K. Cytotoxicity of Tirapazamine (3-Amino-1,2,4-Benzotriazine-1,4-Dioxide)-Induced DNA Damage in Chicken DT40 Cells. Chem. Res. Toxicol. 2017, 30, 699–704.
  95. Guise, C.P.; Mowday, A.M.; Ashoorzadeh, A.; Yuan, R.; Lin, W.-H.; Wu, D.-H.; Smaill, J.B.; Patterson, A.V.; Ding, K. Bioreductive Prodrugs as Cancer Therapeutics: Targeting Tumour Hypoxia. Chin. J. Cancer 2014, 33, 80–86.
  96. Mistry, I.N.; Thomas, M.; Calder, E.D.D.; Conway, S.J.; Hammond, E.M. Clinical Advances of Hypoxia-Activated Prodrugs in Combination With Radiation Therapy. Int. J. Radiat. Oncol. Biol. Phys. 2017, 98, 1183–1196.
  97. O’Connor, L.J.; Cazares-Körner, C.; Saha, J.; Evans, C.N.G.; Stratford, M.R.L.; Hammond, E.M.; Conway, S.J. Design, Synthesis and Evaluation of Molecularly Targeted Hypoxia-Activated Prodrugs. Nat. Protoc. 2016, 11, 781–794.
  98. Stratford, I.J.; Workman, P. Bioreductive Drugs into the next Millennium. Anticancer. Drug Des. 1998, 13, 519–528.
  99. Papandreou, I.; Cairns, R.A.; Fontana, L.; Lim, A.L.; Denko, N.C. HIF-1 Mediates Adaptation to Hypoxia by Actively Downregulating Mitochondrial Oxygen Consumption. Cell Metab. 2006, 3, 187–197.
  100. Cairns, R.A.; Papandreou, I.; Sutphin, P.D.; Denko, N.C. Metabolic Targeting of Hypoxia and HIF1 in Solid Tumours Can Enhance Cytotoxic Chemotherapy. Proc. Natl. Acad. Sci. USA 2007, 104, 9445–9450.
  101. White, E. The Role for Autophagy in Cancer. J. Clin. Investig. 2015, 125, 42–46.
  102. Mizushima, N.; Komatsu, M. Autophagy: Renovation of Cells and Tissues. Cell 2011, 147, 728–741.
  103. Aita, V.M.; Liang, X.H.; Murty, V.V.; Pincus, D.L.; Yu, W.; Cayanis, E.; Kalachikov, S.; Gilliam, T.C.; Levine, B. Cloning and Genomic Organization of Beclin 1, a Candidate Tumour Suppressor Gene on Chromosome 17q21. Genomics 1999, 59, 59–65.
  104. Choi, A.M.K.; Ryter, S.W.; Levine, B. Autophagy in Human Health and Disease. N. Engl. J. Med. 2013, 368, 651–662.
  105. Liang, X.H.; Jackson, S.; Seaman, M.; Brown, K.; Kempkes, B.; Hibshoosh, H.; Levine, B. Induction of Autophagy and Inhibition of Tumourigenesis by Beclin 1. Nature 1999, 402, 672–676.
  106. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The next Generation. Cell 2011, 144, 646–674.
  107. Yun, C.W.; Lee, S.H. The Roles of Autophagy in Cancer. Int. J. Mol. Sci. 2018, 19, 3466.
  108. Rabinowitz, J.D.; White, E. Autophagy and Metabolism. Science 2010, 330, 1344–1348.
  109. Liu, E.Y.; Ryan, K.M. Autophagy and Cancer--Issues We Need to Digest. J. Cell Sci. 2012, 125, 2349–2358.
  110. Verbaanderd, C.; Maes, H.; Schaaf, M.B.; Sukhatme, V.P.; Pantziarka, P.; Sukhatme, V.; Agostinis, P.; Bouche, G. Repurposing Drugs in Oncology (ReDO)—Chloroquine and Hydroxychloroquine as Anti-Cancer Agents. Ecancermedicalscience 2017, 11, 781.
  111. Levy, J.M.M.; Towers, C.G.; Thorburn, A. Targeting Autophagy in Cancer. Nat. Rev. Cancer 2017, 17, 528–542.
  112. Townsend, K.N.; Hughson, L.R.K.; Schlie, K.; Poon, V.I.; Westerback, A.; Lum, J.J. Autophagy Inhibition in Cancer Therapy: Metabolic Considerations for Antitumour Immunity. Immunol. Rev. 2012, 249, 176–194.
  113. Ben-Zvi, I.; Kivity, S.; Langevitz, P.; Shoenfeld, Y. Hydroxychloroquine: From Malaria to Autoimmunity. Clin. Rev. Allergy Immunol. 2012, 42, 145–153.
  114. Viry, E.; Paggetti, J.; Baginska, J.; Mgrditchian, T.; Berchem, G.; Moussay, E.; Janji, B. Autophagy: An Adaptive Metabolic Response to Stress Shaping the Antitumour Immunity. Biochem. Pharmacol. 2014, 92, 31–42.
  115. Cicchini, M.; Karantza, V.; Xia, B. Molecular Pathways: Autophagy in Cancer—A Matter of Timing and Context. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2015, 21, 498–504.
  116. Cheong, H. Integrating Autophagy and Metabolism in Cancer. Arch. Pharm. Res. 2015, 38, 358–371.
  117. Yim, W.W.-Y.; Mizushima, N. Lysosome Biology in Autophagy. Cell Discov. 2020, 6, 1–12.
  118. Tasdemir, E.; Galluzzi, L.; Maiuri, M.C.; Criollo, A.; Vitale, I.; Hangen, E.; Modjtahedi, N.; Kroemer, G. Methods for Assessing Autophagy and Autophagic Cell Death. Methods Mol. Biol. 2008, 445, 29–76.
More